EP1968612B1 - Dinucleotide prodrugs - Google Patents

Dinucleotide prodrugs Download PDF

Info

Publication number
EP1968612B1
EP1968612B1 EP06848625.7A EP06848625A EP1968612B1 EP 1968612 B1 EP1968612 B1 EP 1968612B1 EP 06848625 A EP06848625 A EP 06848625A EP 1968612 B1 EP1968612 B1 EP 1968612B1
Authority
EP
European Patent Office
Prior art keywords
alkyl
aryl
compound
absent
hbv
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP06848625.7A
Other languages
German (de)
English (en)
French (fr)
Other versions
EP1968612A2 (en
EP1968612A4 (en
Inventor
Radhakrishnan P. Iyer
Seetharamaiyer Padmanabhan
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
F Star Therapeutics Inc
Original Assignee
Spring Bank Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Spring Bank Pharmaceuticals Inc filed Critical Spring Bank Pharmaceuticals Inc
Priority to EP16158945.2A priority Critical patent/EP3090748B1/en
Priority to RS20160328A priority patent/RS54867B1/sr
Priority to PL16158945T priority patent/PL3090748T3/pl
Priority to EP19157583.6A priority patent/EP3553072A1/en
Priority to SI200632048A priority patent/SI1968612T1/sl
Priority to DK16158945.2T priority patent/DK3090748T3/da
Publication of EP1968612A2 publication Critical patent/EP1968612A2/en
Publication of EP1968612A4 publication Critical patent/EP1968612A4/en
Application granted granted Critical
Publication of EP1968612B1 publication Critical patent/EP1968612B1/en
Priority to CY20161100388T priority patent/CY1117576T1/el
Priority to HRP20160531TT priority patent/HRP20160531T1/hr
Priority to CY20191100534T priority patent/CY1121639T1/el
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals
    • C07H19/20Purine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7084Compounds having two nucleosides or nucleotides, e.g. nicotinamide-adenine dinucleotide, flavine-adenine dinucleotide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/04Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with deoxyribosyl as saccharide radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J17/00Normal steroids containing carbon, hydrogen, halogen or oxygen, having an oxygen-containing hetero ring not condensed with the cyclopenta(a)hydrophenanthrene skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J43/00Normal steroids having a nitrogen-containing hetero ring spiro-condensed or not condensed with the cyclopenta(a)hydrophenanthrene skeleton
    • C07J43/003Normal steroids having a nitrogen-containing hetero ring spiro-condensed or not condensed with the cyclopenta(a)hydrophenanthrene skeleton not condensed

Definitions

  • the invention was supported, in whole or in part by NIH Grant number 5 UOI Al058270-02/03.
  • the present invention relates to the design, synthesis, and evaluation of prodrug analogs of nucleosides, nucleotides, and oligonucleotides.
  • the compounds, and compositions of the present invention are useful for the treatment of hepatitis B virus (HBV) infections and liver diseases associated with HBV.
  • HBV hepatitis B virus
  • the compounds and combinations can be administered either alone or in combination with other anti- HBV agents.
  • HBV Hepatitis B virus
  • HBV infection leads to chronic persistent hepatitis, fatigue, liver cirrhosis, liver cancer and death.
  • the epidemiology of HBV infection is similar to that of human immunodeficiency virus (HIV). Many HIV carriers are co-infected with HBV. However, HBV is 100 times more infectious than HIV.
  • anti-HBV drugs Although three anti-HBV drugs have been currently approved for clinical use, significant unmet medical need exists due to rapid emergence of resistance, and dose-limiting toxicity associated with therapy.
  • the drugs approved for clinical use includes alpha interferon, a genetically engineered protein, and nucleoside analogs such as lamivudine, and entacavir.
  • Another approved anti-HBV drug is adefovir dipivoxil, which is considered a mononucleotide phosphonate analog.
  • Adefovir or (9-[2-(phosphono-methoxy)ethyl]adenine also referred to as PMEA is disclosed in US patents 5,641,763 and 5,142,051 .
  • the corresponding prodrug, referred to as adefovir dipivoxyl, is clinically approved as an orally acting anti-HBV agent.
  • US Pat 5,444,063 and 5,684,010 disclose the use of enantiomers of beta-D-1,3-dioxolane nucleoside to treat HBV.
  • US Pat. No. 6,881,831 to Iyer et al. discloses compounds comprising two or more deoxyribonucleotide and/or ribonucleotide monomers connected by internucleotide linkages for use in the treatment of HBV.
  • anti-HBV agents claimed include: (1) beta-D-3'azido-2,3-dideoxy 5-fluorocytidine ( Mahmoudian, Pharm Research 8, 1198-203, 1991 ; (2) 2'-beta-D-F-2',3'-dideoxynucleoside analogs, Tsai et al., Biochem Pharmacol. 48,1477-1481, 1994 ; (3) 5-carboximido-, or 5-fluro-2,3 unsaturated or 3'-modified pyrimidine nucleosides.
  • nucleotide analogs have also been claimed to be anti-HBV agents. These include 9[1-phosphonomethoxycyclopropyl)methylguanine], PMCG and its dipivaloxyl prodrug, PMCDG and the trifluoromethyl analog, MCC-478.
  • Cyclic nucleoside phosphonate analogs and prodrug derivatives are also nucleotide analogs with anti-HBV activity.
  • the corresponding phosphoramidate prodrug analogs are converted to the phosphonate derivative by presumably by esterase enzymes.
  • esterase enzymes See: Iyer et al., Current Opinion in Pharmacol., 5, 520-528,2005 .
  • the concept of using chemically modified drugs as prodrug analogs is an established paradigm in the pharmaceutical development of a number of different drugs.
  • the prodrug strategies permit transient modification of the physicochemical properties of the drug in order to: (a) improve chemical stability, (b) alter aqueous solubility, (c) improve bioavailability (d) target specific tissues (e) facilitate synergistic drug combinations, (f) overcome first-pass metabolic effects, (g) serve as lipophilic carrier for hydrophilic drugs, and (h) serve as a chemical depot for sustained drug delivery.
  • a few prodrug strategies have been employed to improve bioavailability, to enhance liver tissue distribution and to improve antiviral potency.
  • modification of the phosphate group as the corresponding amino acid phosphoramidate results in more potent antivirals ( Gudmundsson et al., Nucleosides, Nucleotides, 23, 1929-1937, 2004 . Cahard et al., Mini Reviews Med Chem., 4,371-381, 2004 .
  • Glyceryl phosphate and phospholipid prodrugs of nucleosides have also been developed ( Hostetler et al., Antimicrob Agents and Chemotherapy, 44, 1064-1069, 2000 ) to improve oral bioavailability.
  • S -acylthioethyl (SATE) and cyclic salicyl derivatives (cyclosal) are other examples of prodrug derivativation of nucleosides and nucleotides ( Peyrottes, et al., Mini Rev. Med. Chem., 4, 395-408, 2004 ) and Meier et al., Mini Rev Med Chem 4, 383-394, 2004 .
  • Other prodrug strategies include 4-arylsusbstituted cyclic 1,3-propanyl esters (HepDirect analogs) designed to undergo oxidative cleavage by liver enzymes to release the active nucleotide intracellularly ( Erion et al., J. Am. Chem. Soc., 126, 5154-5163, 2004 ).
  • nucleosides need to be phosphorylated to nucleoside mono-, di-, and triphosphates before they can become inhibitors ofHBV polymerase.
  • nucleosides can be considered as prodrugs, which need to be activated in vivo. Since most nucleosides target viral polymerase and act by similar mechanism of action, there is potential for rapid emergence of resistance and occurrence of adverse events such as mitochondrial toxicity due to inhibition of human gamma polymerase. Another problem with antiviral therapy is viral rebound following cessation of therapy.
  • WO98/07734 discloses an oligonucleotide prodrug comprising at least six covalently linked nucleotides, at least one of which is site-specifically derivatised with a lipophilic chemical group reversibly and covalently attached to the nucleotide at a 5'phosphate, a 3'phosphate or an internucleotidic phosphate linkage.
  • US5,770,713 describes triester phosphorothioate olignucleotides comprising internucleotide concatenations having p-s- bond protected by a bioreversible grouping in intracellular media.
  • Olignucleotides having bioreversible phosphate blocking groups that can be used as prodrugs for the olignucleotide are also described in WO97/476637 .
  • TpT prodrugs The chemical stability of TpT prodrugs was investigated by Poijarvi et al., Nucleosides, Nucleotides & Nucleic acids 20 (1 &2), 77-91 (2001 ).
  • oligonucleotides Being highly charged, large molecular weight compounds, oligonucleotides have unfavorable physicochemical attributes for cell permeation by passive diffusion. Consequently, the design of prodrug analogs of oligonucleotides has mainly focused on the partial masking of some of their negatively charged backbone by bioreversible, lipophilic groups. Several such analogs have been synthesized and bioreversibility has been demonstrated in vitro. However, it appears that although the initial unmasking of one or two nucleotides take place rapidly, complete unmasking takes several hours or even days.
  • lyer et al. prepared S- acyloxyalkyl derivatives of a mixed PO-PS oligonucleotide and found that in vitro, they could convert back to the parent oligonucleotide albeit slowly.
  • a similar SATE prodrug strategy has been employed for oligonucleotide prodrugs. But, there has not been a demonstration of their in vivo potential either in terms of improved pharmacokinetics of oligonucleotides or enhanced biological activity. Also, there are no reports of in vivo oral bioavailability studies of oligonucleotide prodrugs or demonstration of in vivo biological activity.
  • Shorter chain oligonucleotides (less than 8-mers) with lesser number of charges and smaller molecular weight compared to 20-mer oligonucleotides represent a promising class of novel molecules with potential therapeutic and diagnostic properties. Indeed, recent reports suggest that mono-, di-, tri-, and short chain oligonucleotides possess significant biological activity that can be exploited for therapeutic applications.
  • the present invention provides a pronucleotide of formula (III): or the racemates, enantiomers, diastereomers, geometric isomers, tautomers thereof, wherein
  • Prodrugs in accordance with the invention are useful in the prevention and treatment of HBV infections and other conditions caused by HBV such as liver inflammation, liver cirrhosis, acute hepatitis, fulminant hepatitis, chronic hepatitis, and other liver diseases.
  • the compounds and formulations of the invention can also be used prophylactically to prevent disease progression in HBV-infected individuals.
  • a method for the treatment of a HBV infection in a host, including human includes administering an effective amount of a prodrug of the invention including a pharmaceutically active salt thereof, administered alone or in combination or sequentially with another or other anti-HBV agent(s).
  • Preferred prodrugs of the invention comprise di-, and tri-nucleotides including, but not limited to, 3-dApsU 2'-OMe , 3'dApsA 7deaza and 3'-dApsTpsC and their analogs where "ps" refers to phosphorothioate internucleotidic linkages.
  • the compounds of the present invention are compounds represented by formula III illustrated above, or racemates, enantiomers, diastereomers, geometric isomers, tautomers thereof.
  • the compounds of the present invention are compounds represented by formula IV as illustrated below, or racemates, enantiomers, diastereomers, geometric isomers, tautomers thereof.
  • R 4 is selected from hydrogen, C(O)-alkyl, C(O)O-alkyl, C(O)-aryl, C(O)O-aryl, C(O)NH-alkyl, and C(O)NH-aryl; and R, R 3 , X, X 1 , A and n are as previously defined.
  • the compounds of the present invention are compounds represented by formula V as illustrated below, or racemates, enantiomers, diastereomers, geometric isomers, tautomers thereof. wherein R, R 3 , R 4 , X, X 1 , A and n are as previously defined.
  • Representative compounds according to the invention are those selected from the group consisting of: Compounds (1)-(8) of the formula A1: wherein R, X 1 , R 3 and R 4 is delineated for each example in Table 1.
  • TABLE 1 Compound No. R X 1 R 3 R 4 1 absent H H 2 O H H 3 absent H H 4 O H H 5 O C(O)PH H 6 O H C(O)Ph 7 absent H H 8 O H H
  • Representative compounds according to the invention are those selected from the group consisting of: Compounds (9)-(16) of the formula B1: wherein R, X 1 , R 3 and R 4 is delineated for each example in Table 2.
  • TABLE 2 Compound No. R X 1 R 3 R 4 9 absent H H 10 O H H 11 absent H H 12 O H H 13 O C(O)PH H 14 O H C(O)PH 15 absent H H 16 O H H H
  • the lack of oral bioavailability may be due to a number of factors including: (a) the acidic environment in the stomach that cause substantial degradation of the nucleotide, (b) the negative charge on the backbone that suppress permeation of the nucleotide through the intestinal mucosal barrier, and (c) the presence of various digestive enzymes in the GI tract that degrade the compound.
  • both longer and shorter-chain oligonucleotides are not orally bioavailable, it appears that in the case of smaller nucleotide class of compounds, charge rather than the size of the compound may be a more important factor in determining bioavailability and that masking the negative charge on the backbone may potentially provide orally bioavailable di-, and tri-nucleotide compounds.
  • nucleosides in general, are poorly bioavailable orally as such, and prodrug derivatization is adapted as a strategy to enhance oral bioavailability.
  • US Patent 6,875,751 claimed by Imbach et al. reveals 3'-amino acid prodrugs of 2'-deoxy-beta-L-nucleosides as improved orally bioavailable prodrugs of L-nucleosides.
  • SATE prodrug strategy has also been similarly applied for nucleosides.
  • nucleotides and dinucleotides contain highly acid-labile purine and pyrimidine moieties in their structures.
  • masking the negative charge of these molecules might aid in their cellular diffusion by increased lipohilicity, it is not known whether they will be stable in gastric mucosa long enough to be absorbed orally.
  • dinucleotide 3-dApsU 2'-OMe was rapidly degraded in simulated gastric fluid with a half-life less than 10 minutes. Such degradative process is known to occur by initial protonation of nitrogen of the nucleobase followed by depurination and cleavage of the sugar ring.
  • drug molecules should have molecular weight less than 500 daltons, not more than 5 hydrogen bond donors (OH and NH groups) not more than 10 hydrogen bond acceptors (notably N and O), a molecular weight under 500, a LogP under 5 for oral absorption by passive diffusion.
  • hydrogen bond donors OH and NH groups
  • 10 hydrogen bond acceptors notably N and O
  • a molecular weight under 500 a LogP under 5 for oral absorption by passive diffusion.
  • both di-, and trinucleotide prodrugs are higher molecular weight compounds and do not meet many of the Lipinski criteria for oral absorption.
  • di- and trinucleotide prodrugs of the present invention have novel modifications or substitutions in the ring and nucleobases. Since esterases or other enzymes have specific structural and topological requirements for activity, it could not be anticipated whether di- and tri-nucleotide prodrugs would be substrates for the enzymes. In addition, since many compounds described here are isomeric mixtures, and since enzymes known to be stereo-discriminatory, it was not known whether individual isomers could be substrates or whether rate of conversion to the parent molecule would be vastly different that would make them less attractive as drug candidates.
  • prodrugs are known and many strategies exist for making prodrugs of many compounds including nucleosides and mononucleotides, it could not be anticipated a priori, nor is it obvious, for some one skilled in the art, that similar prodrugs of di-, and tri-nucleotides might have oral bioavailability and consequently could be developed as orally bioavailable drugs.
  • the present invention provides such compositions.
  • a number of S -functionalized, uncharged prodrug derivatives are claimed.
  • the design of prodrug derivatives was based upon the ability of a target enzyme to unmask a latent functionality to reveal the parent nucleotide in vivo.
  • the general structure of dinucleotide derivatives 1-3 derived and the expected mechanism for their esterase-mediated conversion to the parent dimer 4 are depicted in Scheme 1, and include: (a) S-(acyloxyalkyl) thiophosphate analogs 1.
  • acyloxyalkyl analogs exemplified by the antibiotics pivampicillin, and bacampicillin, as well as, the recently approved anti-HBV agent adefovir dipivoxil are clinically used, orally bioavailable, ester prodrug analogs. Following their absorption, the conversion of the prodrug to the parent molecule is believed to occur via esterase-mediated hydrolysis in plasma and/or liver, with concomitant liberation of formaldehyde and carboxylic acid. (b) The S -(acyloxyaryl)thiophosphate analogs 2.
  • acyloxyaryl analogs of daunorubicin, doxorubicin, phosphorodiamide mustard, acivicin, and PEG-daunorubicin conjugate are well known 2b and have been extensively evaluated in vitro and in vivo [ Bundgaard, H. In Bio-reversible carriers in drug design. Theory and Application. Roche, E.B. Ed.; Pergamon Press: New York, 1987; pp 13-94 ; For excellent reviews see: Oliyai, R.; Stella, V. J. Annu. Rev Pharmacol. Toxicol.
  • nucleotide analogs or pronucleotides of the invention were designed including the ester analogs (which have a long chain alkoxy group that imparts greater lipophilicity to the molecule), as well as, the amide analog and (c) S -alkyl derivatives with a terminal functional group 3 are designed such that during enzyme- mediated hydrolytic process, a latent nucleophilic group is uncovered, which is juxtapositioned to attack the electrophilic carbon alpha to the thiophosphate moiety resulting in the release of the parent dinucleotide.
  • Prodrugs or pronucleotides of the present invention also relate to certain derivatives and conjugates of nucleotides, dinucleotides, trinucleotides and oligonucleotides.
  • the conjugating moiety can be of different chemical and structural types and can be linked to the hydroxy, amino, phosphate or phosphorothioate backbone of the nucleotides or other functionalities in the nucleoside and oligonucleotides via ester, amide, isocyanate, urea, thiourea, carbamate or other type of covalent linkages.
  • certain conjugates may or may not chemically or enzymatically regenerate the parent nucleotide in vitro or in vivo, and yet the biological activity may reside in the conjugate or in the parent nucleotide or both.
  • several di-, tri-, and tetra-nucleotides and their analogs have been previously identified as anti-HBV agents ( US patent 6,881,83 and US2003/0109471 ).
  • the derivatives and conjugates reported in this invention are applicable to compounds cited in those applications as well.
  • the heterocyclic ring preferably contains 5, or 6-members containing O, N, or S ring atoms free or fused to another ring. Masking the charged backbone may enhance the stability of the nucleotide to the acidic and basic environment of the gastrointestinal tract (having a variety of digestive enzymes) thereby facilitating oral absorption.
  • the presence of a negatively charged phosphoric diester or phosphorothioate linkage is believed to be essential for nuclease-mediated degradation of a polynucleotide.
  • the chemical and enzyme-mediated degradative action of polynucleotide may be inhibited thereby enhancing the stability of the polynucleotide.
  • the conjugating moiety might be a lipophilic group that facilitates the transport of the drug across biological barriers such as the lipid bilayer of mammalian cells or the bacterial cell wall.
  • lipophilic groups include, but not limited to, polyethylene glycol (PEG), cholesterol, cholic acid, phospholipids etc.
  • PEG polyethylene glycol
  • the lipophilic group is linked to either the sugar hydroxyl, the nucleobase, or the internucleotidic phosphate and phosphorothioate linkage at one or more sites as shown in compound (B) illustrating the structure of a cholic acid analog of a dinucleoside phosphorothioate, 3'dApsU 2'OMe or in formula.
  • C-I Typical Structures of dinucleotide conjugated to amino acids, and peptides at the sugar hydroxyls, nucleobase and at the internucleotidic phosphorothioate linkage are represented by compounds (C-I).
  • the conjugating moiety might be a group that facilitates active transport of the nucleotide across various cellular barriers.
  • moieties may be of natural or synthetic origin including amino acids, peptides, and polypeptides.
  • the conjugating moiety may facilitate targeting of the drug to a particular tissue or organ.
  • Such moiety includes monoclonal antibodies or other natural products, which have the property to localize in certain target tissues.
  • a nucleoside unit is represented by the internationally accepted convention of line drawing.
  • a 2'-substituted ribonucleoside is represented in both the conventional structure and the corresponding line drawing format.
  • the sugar units attached to B 1 and B 2 that give rise to ⁇ or ⁇ N- or C- nucleoside includes, but not limited to, furanose, deoxyribofuranose, ribose, and arabinose.
  • aryl refers to a mono- or polycyclic carbocyclic ring system having one or two aromatic rings including, but not limited to, phenyl, naphthyl, tetrahydronaphthyl, indanyl, idenyl and the like.
  • heteroaryl refers to a mono- or polycyclic (e.g. bi-, or tri-cyclic or more) aromatic radical or ring having from five to ten ring atoms of which one or more ring atom is selected from, for example, S, O and N; zero, one or two ring atoms are additional heteroatoms independently selected from, for example, S, O and N; and the remaining ring atoms are carbon, wherein any N or S contained within the ring may be optionally oxidized.
  • Heteroaryl includes, but is not limited to, pyridinyl, pyrazinyl, pyrimidinyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl, isooxazolyl, thiadiazolyl, oxadiazolyl, thiophenyl, furanyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzooxazolyl, quinoxalinyl, and the like.
  • any of the aryls, substituted aryls, heteroaryls and substituted heteroaryls described herein, can be any aromatic group.
  • Aromatic groups can be substituted or unsubstituted.
  • alkyl refers to saturated, straight- or branched-chain hydrocarbon radicals containing between one and six, or one and twelve carbon atoms, respectively.
  • Examples of C 1 -C 6 alkyl radicals include, but are not limited to, methyl, ethyl, propyl, isopropyl, n -butyl, tert -butyl, neopentyl and n-hexyl radicals; and examples of C 1 -C 12 alkyl radicals include, but are not limited to, ethyl, propyl, isopropyl, n-hexyl, octyl, decyl, dodecyl radicals.
  • aralkyl or "arylalkyl” embrace aryl-substituted alkyl radicals such as benzyl, diphenylmethyl, triphenylmethyl, phenylethyl, and diphenylethyl.
  • heterocyclic refers to a non-aromatic 5-, 6- or 7-membered ring or a bi- or tri-cyclic group fused system, where (i) each ring contains between one and three heteroatoms independently selected from oxygen, sulfur and nitrogen, (ii) each 5-membered ring has 0 to 1 double bonds and each 6-membered ring has 0 to 2 double bonds, (iii) the nitrogen and sulfur heteroatoms may optionally be oxidized, (iv) the nitrogen heteroatom may optionally be quaternized, (iv) any of the above rings may be fused to a benzene ring, and (v) the remaining ring atoms are carbon atoms which may be optionally oxo-substituted.
  • heterocycloalkyl groups include, but are not limited to, [1,3]dioxolane, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, piperazinyl, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolidinyl, isothiazolidinyl, quinoxalinyl, pyridazinonyl, and tetrahydrofuryl. Such heterocyclic groups may be further substituted.
  • cycloalkyl denotes a monovalent group derived from a monocyclic or polycyclic saturated carbocyclic ring compound by the removal of a single hydrogen atom. Examples include, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, bicyclo [2.2.1] heptyl, and bicyclo [2.2.2] octyl.
  • substituted aryl refers to aryl, alkyl and cycloalkyl groups as previously defined, substituted by independent replacement of one, two, or three or more of the hydrogen atoms thereon with substituents including, but not limited to, -F, -Cl, -Br, -I, -OH, protected hydroxyl, -NO 2 , -CN, -NH 2 , protected amino, -NH -C 1 -C 12 -alkyl, -NH-C 2 -C 12 -alkenyl, -NH -C 2 -C 12 -alkenyl, -NH -C 3 -C 12 -cycloalkyl, -NH -aryl, -NH - heteroaryl, -NH -heterocycloalkyl, -dialkylamino, -diarylamino, -
  • steroidal refers to any of numerous naturally occurring or synthetic fat-soluble organic compounds having as a basis 17 carbon atoms arranged in four rings and including the sterols and bile acids, adrenal and sex hormones, certain natural drugs such as digitalis compounds, and the precursors of certain vitamins.
  • steroidal structure includes, but not limited to, cholesterol, cholestanol, 3 ⁇ -cyclo-5- ⁇ -cholestan-6- ⁇ -ol, cholic acid, cholesteryl formiate, cholestanyl formiate.
  • modified nucleoside refers to any nucleoside that includes a modified heterocyclic base, a modified sugar moiety, or a combination thereof.
  • the modified nucleoside is a non-natural pyrimidine or purine nucleoside, as herein described.
  • examples of the modified nucleoside include, but not limited to, 2'-substituted ribonucleoside an arabinonucleoside or a 2'-deoxy-2'-flouroarabinoside, deazaadenine, deazaguanine.
  • the R p S p mixture of the phosphorothioate analog 3-dApsU 2'-OMe (5), was synthesized in large scale (I millimol of nucleoside-loaded controlled-pore glass (CPG) support) using solid-phase phosphoramidite chemistry, ( Beaucage, S. L.; lyer, R. P. Tetrahedron 1993, 49, 1925 ) in conjunction with a specially fabricated LOTUS reactor®( Padmanabhan, S.; Coughlin, J. E.; Iyer, R. P. Tetrahedron Lett. 2005, 46, 343 ; Iyer, R. P.; Coughlin, J. E.; Padmanabhan, S.
  • the dA-linked CPG support was prepared using our recently discovered ultrafast functionalization and loading process for solid supports.
  • a solution of 3 H- 1,2-benzodithiole-3-one-1,1,-dioxide (0.4 M in dry CH 3 CN) was employed ( Iyer, R. P.; Regan, J. B.; Egan, W.; Beaucage, S. L. J. Am. Chem. Soc. 1990, 112, 1253 ).
  • prodrugs or pronucleotide derivatives 6a-j were synthesized in yields of 50 to 70% by chemoselective S -alkylation of R p , S p - 5 with the corresponding iodo-, or bromo-derivatives 7a-j in aqueous acetone or methanol, followed by work-up and chromatographic purification.
  • pronucleotides Representative preparation of pronucleotide 6a. To a solution of the dinucleotide sodium salt (50 mg, 0.082 mmol) in water (1 mL) was added a solution of iodomethyl pivalate( 7a (Table 4), 85 mg, 0.35 mmol) in acetone (2 mL). The reaction was stirred overnight in the dark and concentrated with a few mgs of sodium bisulfite. The crude product was purified by column chromatography and 6a eluted out in a mixture of DCM/MeOH(90/10). Concentration in vacuo gave chromatographically pure white solid (31P NMR, 28.7, 27.9 ⁇ ppm). All analogs were prepared using similar procedures (Table 4).
  • the target compound 6k is prepared in two steps.
  • Step 1 Preparation of Iodomethylisopropyl carbonate: To a solution of anhydrous sodium iodide (6 g, 40 mmol ) in anhy. acetonitrile (20 mL) chloromethyl isopropyl carbonate ( 2.9g, 19 mmol) in anhyd. acetonitrile (10 mL) was added dropwise over 20 min. The reaction mixture, covered with aluminum foil (protected from light) was stirred at room temperature overnight. The solid separated was filtered, washed with acetonitrile and the filtrate was concentrated under reduced pressure. Residue was dissolved in water (10 mL) and organics were extracted in ether (25 mL).
  • Step 2 Alkylation of dinucleotide , 3'-ApsU2'OMe.
  • a solution of dinucleotide 60 mg, 0. 098mmol
  • water HPLC, 400 mL
  • acetone 1 mL
  • Additional acetone (1 mL) was added to get a clear solution to avoid any separation of oily globules of alkylating agent.
  • the reaction mixture covered in aluminum foil, was stirred for 3 h, concentrated under rotavap conditions and later in high vacuum to obtain the reaction mixture as a white solid.
  • Step 1 Synthesis of chloromethyl deoxycholate.
  • deoxycholic acid 120 mg, 0.306 mmol
  • ethanol 4 mL
  • caesium carbonate 53 mg, 0.160 mmol
  • water 3 mL
  • the reaction mixture was stirred for 30 min and ethanol was initially removed under rotavap, and later under high vac. The residue was lyophilized to give the cesium salt as white powder.
  • N- dimethylformamide (DMF, 3 mL) at room temperature bromochloromethane (10 mL) was added and the aluminum foil covered reaction mixture was stirred at room temperature for 24 h.
  • DMF N- dimethylformamide
  • Step 2 Preparation of iodomethyl deoxycholate To a solution of sodium iodide (304 mg, 2.03 mmol) in anhyd. acetonitrile (3mL) chloromethyl ester (438 mg, 0.99 mmol) in a mixture of acetonitrile (6 mL) and dichloromethane (2 mL) was added slowly. The reaction mixture, protection from light, was stirred at room temperature over 48 hours. After concentration, the reaction mixture was extracted in dichloromethane (15 mL), organic layer was washed with water (5 mL), sodium bisulfite (5%, 5 mL) and finally brined (5 mL). Dried over anhyd. sodium sulfate and the crude product, obtained after removal of solvent, was purified by silica column chromatography to obtain the iodo compound (110 mg, 21%).
  • Step 3 Coupling of iodomethyl deoxycholate.
  • 3'dApsU2'OMe 50 mg, 0.082 mmol
  • acetone 3 mL
  • iodomethyl deoxycholate 110 mg, 2.066 mmol
  • acetone 3 mL
  • the solid separated was dissolved by adding more acetone ( ⁇ 6 mL) and the reaction mixture ) was stirred overnight.
  • Step 1 Preparation of 4-acetoxybenzyl alcohol: To a cooled suspension of 4-hydroxybenzyl alcohol (1.95g, 14 mmol) in ethyl acetate (25 mL) in an ice-bath, triethylamine (2.1 mL, 14.9 mmol) was added in one lot under stirring. A solution of acetyl chloride (1.1 mL, 15.5 mmol) in ethyl acetate (12 mL) was added dropwise from an addition funnel. The reaction mixture was stirred overnight.
  • Step 2 Preparation of 4-acetoxybenzyl iodide: To a solution of 4-acetoxybenzyl alcohol (0.332 g, 2 mmol), and cesium iodide (0.571 g, 2.2 mmol) in anhyd. acetonitrile (10 mL) under nitrogen, boron trifluoride etherate (0.28 mL, 2.2 mmol) in acetonitrile (5 mL) was introduced. After stirring overnight, the reaction mixture was poured into ice-cold water (20 mL) and the solid separated was filtered, washed with water and later with hexanes. The product was dried under high vacuum.
  • Step 3 Synthesis of 4-acetoxybenzyl analog of 3'dApsU 2'OMe . Alkylation of 3'dApsU 2'OMe with 4-acetoxybenzyl iodide was carried out as before.
  • EXAMPLE 9 Cytotoxicity Assays: Standard MTT assays were performed in 96-well plates using the Promega CellTiter96 Non-radioactive Cell Proliferation Assay Kit in conjunction with a 96-well Plate Reader (ThermoMax,Molecular devices), and using MDBK, Vero, and HFF cell lines (obtained from ATCC).
  • Bioreversibility studies were carried out as follows: A stock solution of each analog was prepared by dissolving 2 mg in100 ⁇ L of DMSO. 10 ⁇ L aliquots were diluted with 90 ⁇ L of phosphate buffer (0.1 M, pH 7.0) and 100 ⁇ L aliquots of rabbit serum. The mixture was incubated at 37 oC in a water bath. Aliquots were removed at different time points and diluted with200 ⁇ L of methanol to stop the reaction. The incubate was then centrifuged, supernatant concentrated in a speed vac and diluted with 200 ⁇ L of 0.1 M ammonium acetate buffer prior to injection into HPLC.
  • phosphate buffer 0.1 M, pH 7.0
  • Reversed-phase HPLC analysis was carried out using a Waters Instrument equipped with a 600Egradient controller, and a 996 photodiode array detector with Millennium software.
  • X-terra MS C18 2.5 ⁇ m, 2.1 X 20 mm column and an operating gradient of 100% A to 80% B over 30 minutes of buffer A (0.1 M NH4OAc) and buffer B (80:20,CH3CN:NH4OAc) was employed.
  • Retention time for prodrugs ranged from 16 to 18 minutes whereas that of the R p, S p dinucleotide 5 was 13. 5, 13.8 minutes.
  • amino acid derived prodrug 6m and carbonate derivative 6k underwent almost complete conversion to 3'dApsU 2'OMe in ⁇ 3 h on serum treatment.
  • Other prodrugs had different rates of conversion to the parent dinucleotide. Some prodrugs did not convert back to the parent under the conditions of the experiment.
  • EXAMPLE 11 Stability. The stability of prodrugs in simulated gastric fluid (SGF) and simulated intestine fluid (SIF) at 37 °C were examined. SGF and SIF were prepared following reported procedures and prodrugs were incubated separately with SGF and SIF for 1 h at 37 °C, processed, and analyzed using reverse-phase HPLC. It was found that parent dinucleotide 3''dApsU 2'OMe was not stable in SGF decomposing in about 15 min but was relatively stable in SIF. All prodrugs were significantly stable in SGF with half-lives ranging from 1 to 3 hours. In SIF, S-acyloxyalkyl prodrugs were converted to the parent dinucleotide with half-life of about 1 h.
  • SGF gastric fluid
  • SIF simulated intestine fluid
  • EXAMPLE 12 Oral bioavailability.
  • the oral bioavailability of the prodrugs was determined in CD-1 mice. Each of the representative prodrugs 6a, 6k, 6l were dissolved in water and administered to groups of mice by oral gavage. Male Swiss-Webster mice weighing between 20 to 30 g (Charles River Labs) were used for the study. At designated time points of 5, 15, 30, 60 and 120 minutes, mice were sacrificed and the blood collected by cardiac puncture. Liver, kidney, stomach, duodenum, jejunum, ileum and brain were removed and frozen in dry ice until processing. Plasma was separated from blood by centrifugation and processed for analysis of the drug content by reversed-phase HPLC.
  • Levels of each prodrug and/or the parent 3'dApsU 2'OMe were determined by analytical HPLC. Tissue samples (principally liver) were processed following homogenization in 1% SDS in the presence of 0.1 M NaOAc. The homogenate was added to a PALL 50K concentrator and centrifuged for 2 h at 3000 rpm. A sample was run on a reversed-phase HPLC column (2.1X20 mm X-Terra column), flow rate 1ml/min, 30 min gradient of 100% A (0.1M NH 4 OAc) to 100% B (acetonitrile: 0.1 M NH 4 OAC, 80:20).
  • prodrug could be detected in early time points whereas at later time points, mainly the parent dinucleotide 3'dApsU 2'OMe was seen. In the case of liver, mainly 3'dApsU 2'OMe was seen. These observations are consistent with oral absorption of the prodrug followed by enzyme-mediated conversion of the prodrug to the 3'dApsU 2'OMe . Most likely, the enzymes responsible for the conversion of the prodrug to 3'dApsU24O 2'OMe are esterases found both in blood and in tissues. The estimate of oral bioavailability ranges from 5 to 15% in plasma and liver.
  • EXAMPLE 13 In vivo anti-HBV activity of the prodrugs. Certain prodrugs were evaluated in the transgenic mouse model of HBV infection. Male transgenic mouse infected with HBV, with age ranging from 78 to 108 days were used. The prodrugs 6a and 6k were initially evaluated at a single dose of 300 to 400 mg/kg, administered daily for 14 days by oral gavage. The compounds were administered in citric acid and adefovir dipivoxil was used as a positive control. A control group which received vehicle was used a negative control. Following the treatment, mice were sacrificed and liver tissue analyzed for HBV DNA using Southern blot analysis. The data was statistically evaluated using Kruskall-Wallis non-parametric ANOVA and are the plot shown in Figure 2 . Both prodrugs 6a and 6k produced up to 2 log reduction of liver HBV DNA compared to untreated control, which was statistically significant with p values 0.01 to 0.001.
  • mice Male and female transgenic mice (founder 1.3.32) were infected with human hepatitis B virus. Following infection, animals were orally administered compounds 6a or 6k , or a placebo of 0.05 M citric acid, pH 2.0 once daily for 14 days. Dosages were 400 mg/kg/d, for compound 6a and 300 mg/kg/d for compound 6k . The positive control, ADV, was administered at 10 mg/kg/d. The data are summarized in Tables 6 and 7. Statistical significance is indicated as *P ⁇ 0.05, **P ⁇ 0.01, ***P ⁇ 0.001 compared to placebo vehicle. Measurements of serum HBeAg, PEI are reported according to International Immuno Diagnostics standardized assay using Paul Ehrlich International Units (PEI U).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Virology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Saccharide Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Steroid Compounds (AREA)
EP06848625.7A 2005-12-13 2006-12-13 Dinucleotide prodrugs Active EP1968612B1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
EP16158945.2A EP3090748B1 (en) 2005-12-13 2006-12-13 Compositions comprising nucleotide and oligonucleotide prodrugs
RS20160328A RS54867B1 (sr) 2005-12-13 2006-12-13 Dinukleotidni prolekovi
PL16158945T PL3090748T3 (pl) 2005-12-13 2006-12-13 Kompozycje zawierające proleki nukleotydowe i oligonukleotydowe
EP19157583.6A EP3553072A1 (en) 2005-12-13 2006-12-13 Compositions comprising dinucleotide prodrugs
SI200632048A SI1968612T1 (sl) 2005-12-13 2006-12-13 Dinukleotid predzdravila
DK16158945.2T DK3090748T3 (da) 2005-12-13 2006-12-13 Sammensætninger, der omfatter nukleotid- og oligonukleotid-prodrugs
CY20161100388T CY1117576T1 (el) 2005-12-13 2016-05-09 Δινουκλεοτιδικα προφαρμακα
HRP20160531TT HRP20160531T1 (hr) 2005-12-13 2016-05-18 Dinukleotidni prolijekovi
CY20191100534T CY1121639T1 (el) 2005-12-13 2019-05-20 Συνθεσεις που περιλαμβανουν προφαρμακα νουκλεοτιδιων και ολιγονουκλεοτιδιων

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US75003605P 2005-12-13 2005-12-13
US80029406P 2006-05-15 2006-05-15
US11/637,520 US8076303B2 (en) 2005-12-13 2006-12-12 Nucleotide and oligonucleotide prodrugs
PCT/US2006/047617 WO2007070598A2 (en) 2005-12-13 2006-12-13 Nucleotide and oligonucleotide prodrugs

Related Child Applications (2)

Application Number Title Priority Date Filing Date
EP16158945.2A Division EP3090748B1 (en) 2005-12-13 2006-12-13 Compositions comprising nucleotide and oligonucleotide prodrugs
EP19157583.6A Division EP3553072A1 (en) 2005-12-13 2006-12-13 Compositions comprising dinucleotide prodrugs

Publications (3)

Publication Number Publication Date
EP1968612A2 EP1968612A2 (en) 2008-09-17
EP1968612A4 EP1968612A4 (en) 2013-01-09
EP1968612B1 true EP1968612B1 (en) 2016-03-09

Family

ID=38194676

Family Applications (3)

Application Number Title Priority Date Filing Date
EP06848625.7A Active EP1968612B1 (en) 2005-12-13 2006-12-13 Dinucleotide prodrugs
EP19157583.6A Withdrawn EP3553072A1 (en) 2005-12-13 2006-12-13 Compositions comprising dinucleotide prodrugs
EP16158945.2A Active EP3090748B1 (en) 2005-12-13 2006-12-13 Compositions comprising nucleotide and oligonucleotide prodrugs

Family Applications After (2)

Application Number Title Priority Date Filing Date
EP19157583.6A Withdrawn EP3553072A1 (en) 2005-12-13 2006-12-13 Compositions comprising dinucleotide prodrugs
EP16158945.2A Active EP3090748B1 (en) 2005-12-13 2006-12-13 Compositions comprising nucleotide and oligonucleotide prodrugs

Country Status (17)

Country Link
US (6) US8076303B2 (el)
EP (3) EP1968612B1 (el)
JP (1) JP5044854B2 (el)
KR (2) KR101248873B1 (el)
CN (2) CN101437397B (el)
CY (2) CY1117576T1 (el)
DK (2) DK3090748T3 (el)
ES (2) ES2573928T3 (el)
HR (1) HRP20160531T1 (el)
HU (2) HUE029190T2 (el)
IN (1) IN2015DN01786A (el)
LT (1) LT3090748T (el)
PL (2) PL3090748T3 (el)
PT (1) PT3090748T (el)
RS (1) RS54867B1 (el)
SI (2) SI1968612T1 (el)
WO (1) WO2007070598A2 (el)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10246480B2 (en) 2017-02-17 2019-04-02 Eisai R&D Management Co., Ltd. Compounds for the treatment of cancer
US11691990B2 (en) 2018-08-16 2023-07-04 Eisai R&D Management Co., Ltd Salts of compounds and crystals thereof

Families Citing this family (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4012070B2 (ja) * 2001-01-16 2007-11-21 カン−フィテ・バイオファーマ・リミテッド ウイルスの複製を阻害するためのアデノシンa3受容体アゴニストの使用
US8076303B2 (en) * 2005-12-13 2011-12-13 Spring Bank Pharmaceuticals, Inc. Nucleotide and oligonucleotide prodrugs
GB2471806B (en) 2008-04-03 2012-12-19 Spring Bank Pharmaceuticals Inc Compositions and methods for treating viral infections
EP3067359A1 (en) * 2008-09-23 2016-09-14 Scott G. Petersen Self delivering bio-labile phosphate protected pro-oligos for oligonucleotide based therapeutics and mediating rna interference
CA2744987C (en) 2008-12-02 2018-01-16 Chiralgen, Ltd. Method for the synthesis of phosphorus atom modified nucleic acids
JP2012513450A (ja) * 2008-12-23 2012-06-14 ギリンダス・アメリカ・インコーポレイテッド 硫化剤およびオリゴヌクレオチドを合成するためのその使用
BR112012000828A8 (pt) 2009-07-06 2017-10-10 Ontorii Inc Novas pró-drogas de ácido nucleico e métodos de uso das mesmas
AU2015255202B2 (en) * 2009-07-06 2017-07-27 Wave Life Sciences Ltd. Novel nucleic acid prodrugs and methods of use thereof
CN101659612B (zh) * 2009-09-24 2013-01-09 华润赛科药业有限责任公司 一种选择性酯化的方法
EP2611451A4 (en) * 2010-08-30 2014-01-15 Spring Bank Pharmaceuticals Inc DESIGN OF OLIGONUCLEOTIDE ANALOGUES AS THERAPEUTIC AGENTS
WO2012039448A1 (ja) 2010-09-24 2012-03-29 株式会社キラルジェン 不斉補助基
US9605019B2 (en) 2011-07-19 2017-03-28 Wave Life Sciences Ltd. Methods for the synthesis of functionalized nucleic acids
KR101327026B1 (ko) 2011-11-29 2013-11-13 현대자동차주식회사 운전자 보호장치
AU2013288048A1 (en) 2012-07-13 2015-01-22 Wave Life Sciences Ltd. Asymmetric auxiliary group
KR101835401B1 (ko) 2012-07-13 2018-03-08 신 니뽄 바이오메디칼 라보라토리즈, 엘티디. 키랄 핵산 어쥬번트
KR20220139425A (ko) 2012-07-13 2022-10-14 웨이브 라이프 사이언시스 리미티드 키랄 제어
WO2014127378A2 (en) * 2013-02-18 2014-08-21 Spring Bank Pharmaceuticals, Inc. Design of short oligonucleotides as vaccine adjuvants and therapeutic agents
EP3095461A4 (en) 2014-01-15 2017-08-23 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having immunity induction activity, and immunity induction activator
US10322173B2 (en) 2014-01-15 2019-06-18 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having anti-allergic activity, and anti-allergic agent
WO2015108048A1 (ja) 2014-01-15 2015-07-23 株式会社新日本科学 抗腫瘍作用を有するキラル核酸アジュバンド及び抗腫瘍剤
PT3094728T (pt) 2014-01-16 2022-05-19 Wave Life Sciences Ltd Desenho quiral
GB201403697D0 (en) * 2014-03-03 2014-04-16 Link Technologies Ltd Compounds and methods of use
CN106794188B (zh) * 2014-09-28 2022-12-02 华辉安健(北京)生物科技有限公司 聚合胆汁酸衍生物抑制乙型肝炎病毒和丁型肝炎病毒和ntcp运输
US20190046552A1 (en) * 2015-04-07 2019-02-14 Spring Bank Pharmaceuticals, Inc. Compositions and methods for the treatment of hcv infection
AU2016287580A1 (en) * 2015-07-02 2018-02-15 Spring Bank Pharmaceuticals, Inc. Compositions and methods for the treatment of viral infection
WO2017156391A1 (en) * 2016-03-11 2017-09-14 Spring Bank Pharmaceuticals, Inc. Compounds and compositions for the treatment of infections
EP3484504A4 (en) * 2016-07-15 2020-07-29 Sperovie Biosciences, Inc. COMPOUNDS, COMPOSITIONS AND METHODS OF TREATMENT OF DISEASE
JOP20170192A1 (ar) * 2016-12-01 2019-01-30 Takeda Pharmaceuticals Co داي نوكليوتيد حلقي
AR113224A1 (es) 2017-04-28 2020-02-19 Novartis Ag Conjugados de anticuerpo que comprenden un agonista de sting
US11707531B2 (en) * 2017-09-11 2023-07-25 F-star Therapeutics, Inc. Compounds, compositions, and methods for the treatment of disease
WO2019051488A1 (en) 2017-09-11 2019-03-14 Sperovie Biosciences, Inc. COMPOUNDS, COMPOSITIONS AND METHODS OF TREATING DISEASE
CN110467646A (zh) * 2018-05-09 2019-11-19 博瑞生物医药(苏州)股份有限公司 双核苷酸前体药物
CN110467647B (zh) * 2018-05-09 2022-08-30 博瑞生物医药(苏州)股份有限公司 双核苷酸前体药物的制备方法
WO2019219070A1 (zh) * 2018-05-18 2019-11-21 正大天晴药业集团股份有限公司 氘代的低聚核苷酸及前体药物
JP7335277B2 (ja) 2018-06-01 2023-08-29 エーザイ・アール・アンド・ディー・マネジメント株式会社 膀胱がんの治療のための方法
FR3082434B1 (fr) 2018-06-14 2021-04-30 Cairdac Implant cardiaque autonome de type "capsule leadless", comprenant un recuperateur d'energie a lame piezoelectrique
WO2020089815A1 (en) 2018-10-31 2020-05-07 Novartis Ag Antibody conjugates comprising sting agonist
EP3873938A1 (en) 2018-10-31 2021-09-08 Novartis AG Dc-sign antibody conjugates comprising sting agonists
WO2020114495A1 (zh) * 2018-12-06 2020-06-11 正大天晴药业集团股份有限公司 二核苷酸化合物及其前体药物
CN111484541B (zh) * 2019-01-25 2023-06-02 博瑞生物医药(苏州)股份有限公司 双核苷酸前体药物及其制备方法
CN111484540B (zh) * 2019-01-25 2023-09-08 博瑞生物医药(苏州)股份有限公司 含双核苷酸结构的化合物
JP7421280B2 (ja) 2019-07-30 2024-01-24 三和シヤッター工業株式会社 建具

Family Cites Families (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US688183A (en) 1900-12-17 1901-12-03 Carl C Lantz Adjusting-clip for garment-supporters.
DE2817041A1 (de) 1978-04-19 1979-10-31 Reich Maschf Gmbh Karl Verfahren und vorrichtung zum aufteilen von platten
CS264222B1 (en) 1986-07-18 1989-06-13 Holy Antonin N-phosphonylmethoxyalkylderivatives of bases of pytimidine and purine and method of use them
US5204466A (en) 1990-02-01 1993-04-20 Emory University Method and compositions for the synthesis of bch-189 and related compounds
US5914331A (en) 1990-02-01 1999-06-22 Emory University Antiviral activity and resolution of 2-hydroxymethyl-5-(5-fluorocytosin-1-yl)-1,3-oxathiolane
ES2147721T3 (es) 1990-04-18 2000-10-01 Procter & Gamble Pharma Quinolonil lactamas antimicrobianas.
US5444063A (en) 1990-12-05 1995-08-22 Emory University Enantiomerically pure β-D-dioxolane nucleosides with selective anti-Hepatitis B virus activity
US5432165A (en) 1992-04-06 1995-07-11 Oclassen Pharmaceuticals, Inc. Methods for the treatment of infection caused by Hepatitis B virus (HBV)
US5955591A (en) * 1993-05-12 1999-09-21 Imbach; Jean-Louis Phosphotriester oligonucleotides, amidites and method of preparation
FR2705099B1 (fr) * 1993-05-12 1995-08-04 Centre Nat Rech Scient Oligonucléotides phosphorothioates triesters et procédé de préparation.
FR2709754B1 (fr) 1993-09-10 1995-12-01 Centre Nat Rech Scient Composés 2' ou 3'-déoxy- et 2', 3'-didéoxy-beta-L-pentofuranonucléosides, procédé de préparation et application thérapeutique, notamment anti-virale.
US5587362A (en) 1994-01-28 1996-12-24 Univ. Of Ga Research Foundation L-nucleosides
US5581639A (en) 1995-05-04 1996-12-03 National Research Council Of Canada Raman-nath diffraction grating
AU4156197A (en) 1996-08-21 1998-03-06 Hybridon, Inc. Oligonucleotide prodrugs
AU4988697A (en) 1996-10-24 1998-05-15 Vion Pharmaceuticals, Inc. Monophosphate prodrugs of beta-l-fd4c and beta-l-fddc as potent antiviral agents
US6444652B1 (en) 1998-08-10 2002-09-03 Novirio Pharmaceuticals Limited β-L-2'-deoxy-nucleosides for the treatment of hepatitis B
ES2579903T3 (es) 1998-08-10 2016-08-17 Centre National De La Recherche Scientifique (Cnrs) Beta-L-2'-desoxi-nucleósidos para el tratamiento de la hepatitis B
CA2599597A1 (en) 1998-08-10 2000-02-24 Idenix (Cayman) Limited .beta.-l-2'-deoxy-nucleosides for the treatment of hepatitis b
US6875751B2 (en) 2000-06-15 2005-04-05 Idenix Pharmaceuticals, Inc. 3′-prodrugs of 2′-deoxy-β-L-nucleosides
JP4651264B2 (ja) 2000-07-21 2011-03-16 ギリアード サイエンシーズ, インコーポレイテッド ホスホネートヌクレオチドアナログのプロドラッグならびにこれを選択および作製するための方法。
CN101113160A (zh) 2001-05-16 2008-01-30 米珍尼克斯公司 基于核酸的化合物及其使用方法
US8076303B2 (en) * 2005-12-13 2011-12-13 Spring Bank Pharmaceuticals, Inc. Nucleotide and oligonucleotide prodrugs
CN101146337B (zh) 2006-09-15 2011-04-20 华为技术有限公司 新接入节点随机接入的方法及其系统
GB2471806B (en) * 2008-04-03 2012-12-19 Spring Bank Pharmaceuticals Inc Compositions and methods for treating viral infections

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10246480B2 (en) 2017-02-17 2019-04-02 Eisai R&D Management Co., Ltd. Compounds for the treatment of cancer
US10618930B2 (en) 2017-02-17 2020-04-14 Eisai R&D Management Co., Ltd. Compounds for the treatment of cancer
US11339188B2 (en) 2017-02-17 2022-05-24 Eisai R&D Management Co., Ltd. Compounds for the treatment of cancer
US11691990B2 (en) 2018-08-16 2023-07-04 Eisai R&D Management Co., Ltd Salts of compounds and crystals thereof

Also Published As

Publication number Publication date
EP1968612A2 (en) 2008-09-17
PL1968612T3 (pl) 2016-11-30
CN101437397A (zh) 2009-05-20
CY1117576T1 (el) 2017-04-26
ES2729657T3 (es) 2019-11-05
EP3090748B1 (en) 2019-02-20
LT3090748T (lt) 2019-08-26
KR20080072084A (ko) 2008-08-05
US10047114B2 (en) 2018-08-14
SI1968612T1 (sl) 2016-10-28
US10344046B2 (en) 2019-07-09
IN2015DN01786A (el) 2015-07-10
DK3090748T3 (da) 2019-05-27
WO2007070598A3 (en) 2008-10-30
US8076303B2 (en) 2011-12-13
CN102796155A (zh) 2012-11-28
US20120264709A1 (en) 2012-10-18
US20200087338A1 (en) 2020-03-19
US20070149462A1 (en) 2007-06-28
CN102796155B (zh) 2015-08-26
PT3090748T (pt) 2019-06-04
EP1968612A4 (en) 2013-01-09
WO2007070598A8 (en) 2008-12-31
DK1968612T3 (en) 2017-05-22
JP2009519345A (ja) 2009-05-14
WO2007070598A9 (en) 2007-11-08
PL3090748T3 (pl) 2020-03-31
HUE044034T2 (hu) 2019-09-30
HUE029190T2 (en) 2017-02-28
SI3090748T1 (sl) 2019-09-30
EP3090748A1 (en) 2016-11-09
HRP20160531T1 (hr) 2016-08-12
KR101248873B1 (ko) 2013-04-02
WO2007070598A2 (en) 2007-06-21
US20180273574A1 (en) 2018-09-27
ES2573928T3 (es) 2016-06-13
US20140323554A1 (en) 2014-10-30
US20160060287A1 (en) 2016-03-03
CN101437397B (zh) 2012-09-05
RS54867B1 (sr) 2016-10-31
JP5044854B2 (ja) 2012-10-10
EP3553072A1 (en) 2019-10-16
CY1121639T1 (el) 2020-07-31
KR101123534B1 (ko) 2012-03-13
KR20110125677A (ko) 2011-11-21
US8691787B2 (en) 2014-04-08

Similar Documents

Publication Publication Date Title
EP1968612B1 (en) Dinucleotide prodrugs
AU783142B2 (en) Nucleoside analogs with carboxamidine modified monocyclic base
US6815542B2 (en) Nucleoside compounds and uses thereof
US5693769A (en) Glycosylated steroid derivatives for transport across biological membranes and process for making and using same
NZ249715A (en) 2',5'-oligoadenylate derivatives; compounds, pharmaceutical compositions and conjugates
ZA200206468B (en) Nucleoside analogs with carboxamidine modified monocyclic base.

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20080714

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK RS

R17D Deferred search report published (corrected)

Effective date: 20081030

RIC1 Information provided on ipc code assigned before grant

Ipc: C07H 19/04 20060101ALI20081111BHEP

Ipc: C07H 19/00 20060101ALI20081111BHEP

Ipc: A61K 31/70 20060101ALI20081111BHEP

Ipc: A01N 43/04 20060101AFI20081111BHEP

Ipc: C07H 21/00 20060101ALI20081111BHEP

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1122469

Country of ref document: HK

REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Ref document number: 602006048219

Country of ref document: DE

Free format text: PREVIOUS MAIN CLASS: A61K0031707600

Ipc: A61K0031700000

A4 Supplementary search report drawn up and despatched

Effective date: 20121210

RIC1 Information provided on ipc code assigned before grant

Ipc: C07H 21/00 20060101ALI20121204BHEP

Ipc: A61P 31/20 20060101ALI20121204BHEP

Ipc: C07H 19/04 20060101ALI20121204BHEP

Ipc: A61K 31/70 20060101AFI20121204BHEP

Ipc: C07H 19/00 20060101ALI20121204BHEP

17Q First examination report despatched

Effective date: 20131212

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20150522

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: SPRING BANK PHARMACEUTICALS, INC.

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK RS

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 778981

Country of ref document: AT

Kind code of ref document: T

Effective date: 20160315

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602006048219

Country of ref document: DE

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: HR

Ref legal event code: TUEP

Ref document number: P20160531

Country of ref document: HR

REG Reference to a national code

Ref country code: PT

Ref legal event code: SC4A

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20160518

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2573928

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20160613

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: FIAMMENGHI-FIAMMENGHI, CH

REG Reference to a national code

Ref country code: RO

Ref legal event code: EPE

REG Reference to a national code

Ref country code: HR

Ref legal event code: T1PR

Ref document number: P20160531

Country of ref document: HR

REG Reference to a national code

Ref country code: EE

Ref legal event code: FG4A

Ref document number: E012016

Country of ref document: EE

Effective date: 20160601

REG Reference to a national code

Ref country code: GR

Ref legal event code: EP

Ref document number: 20160401164

Country of ref document: GR

Effective date: 20160729

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602006048219

Country of ref document: DE

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 11

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160309

REG Reference to a national code

Ref country code: SK

Ref legal event code: T3

Ref document number: E 22308

Country of ref document: SK

26N No opposition filed

Effective date: 20161212

REG Reference to a national code

Ref country code: HU

Ref legal event code: AG4A

Ref document number: E029190

Country of ref document: HU

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20170516

Ref country code: DK

Ref legal event code: EGE

Effective date: 20170516

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 12

REG Reference to a national code

Ref country code: HK

Ref legal event code: GR

Ref document number: 1122469

Country of ref document: HK

REG Reference to a national code

Ref country code: AT

Ref legal event code: UEP

Ref document number: 778981

Country of ref document: AT

Kind code of ref document: T

Effective date: 20160309

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20160531

Country of ref document: HR

Payment date: 20181121

Year of fee payment: 13

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20160531

Country of ref document: HR

Payment date: 20191204

Year of fee payment: 14

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: HU

Payment date: 20191216

Year of fee payment: 14

Ref country code: FI

Payment date: 20191227

Year of fee payment: 14

Ref country code: MC

Payment date: 20191205

Year of fee payment: 14

Ref country code: CZ

Payment date: 20191204

Year of fee payment: 14

Ref country code: LT

Payment date: 20191203

Year of fee payment: 14

Ref country code: SE

Payment date: 20191227

Year of fee payment: 14

Ref country code: NL

Payment date: 20191226

Year of fee payment: 14

Ref country code: IE

Payment date: 20191227

Year of fee payment: 14

Ref country code: RO

Payment date: 20191206

Year of fee payment: 14

Ref country code: PT

Payment date: 20191205

Year of fee payment: 14

Ref country code: SK

Payment date: 20191204

Year of fee payment: 14

Ref country code: BG

Payment date: 20191218

Year of fee payment: 14

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: PL

Payment date: 20191210

Year of fee payment: 14

Ref country code: GR

Payment date: 20191230

Year of fee payment: 14

Ref country code: FR

Payment date: 20191226

Year of fee payment: 14

Ref country code: IT

Payment date: 20191219

Year of fee payment: 14

Ref country code: LV

Payment date: 20191206

Year of fee payment: 14

Ref country code: IS

Payment date: 20191205

Year of fee payment: 14

Ref country code: EE

Payment date: 20191205

Year of fee payment: 14

Ref country code: LU

Payment date: 20191227

Year of fee payment: 14

Ref country code: SI

Payment date: 20191204

Year of fee payment: 14

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: TR

Payment date: 20191209

Year of fee payment: 14

Ref country code: AT

Payment date: 20191204

Year of fee payment: 14

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DK

Payment date: 20191230

Year of fee payment: 14

Ref country code: GB

Payment date: 20200102

Year of fee payment: 14

Ref country code: ES

Payment date: 20200102

Year of fee payment: 14

Ref country code: DE

Payment date: 20191231

Year of fee payment: 14

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20191231

Year of fee payment: 14

Ref country code: BE

Payment date: 20191227

Year of fee payment: 14

Ref country code: CY

Payment date: 20191206

Year of fee payment: 14

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602006048219

Country of ref document: DE

REG Reference to a national code

Ref country code: HR

Ref legal event code: PBON

Ref document number: P20160531

Country of ref document: HR

Effective date: 20201213

REG Reference to a national code

Ref country code: FI

Ref legal event code: MAE

REG Reference to a national code

Ref country code: EE

Ref legal event code: MM4A

Ref document number: E012016

Country of ref document: EE

Effective date: 20201231

REG Reference to a national code

Ref country code: DK

Ref legal event code: EBP

Effective date: 20201231

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201213

Ref country code: CZ

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201213

Ref country code: CY

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201213

Ref country code: PT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210614

Ref country code: RO

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201213

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: SE

Ref legal event code: EUG

REG Reference to a national code

Ref country code: NL

Ref legal event code: MM

Effective date: 20210101

REG Reference to a national code

Ref country code: AT

Ref legal event code: MM01

Ref document number: 778981

Country of ref document: AT

Kind code of ref document: T

Effective date: 20201213

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20201213

REG Reference to a national code

Ref country code: SK

Ref legal event code: MM4A

Ref document number: E 22308

Country of ref document: SK

Effective date: 20201213

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LV

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201213

Ref country code: MC

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210104

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20201231

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210101

REG Reference to a national code

Ref country code: LT

Ref legal event code: MM4D

Effective date: 20201213

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BG

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210630

Ref country code: AT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201213

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201213

Ref country code: HU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201214

Ref country code: IT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201213

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201213

Ref country code: EE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201231

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201231

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201214

Ref country code: GR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210707

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201231

Ref country code: SK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201213

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201231

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210701

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201213

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201213

Ref country code: DK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201231

REG Reference to a national code

Ref country code: SI

Ref legal event code: KO00

Effective date: 20211203

REG Reference to a national code

Ref country code: ES

Ref legal event code: FD2A

Effective date: 20220214

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201214

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201214

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201213

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201231

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201213